SwePub
Sök i SwePub databas

  Extended search

Träfflista för sökning "WFRF:(Simonsson Ulrika S.H.) "

Search: WFRF:(Simonsson Ulrika S.H.)

  • Result 1-10 of 113
Sort/group result
   
EnumerationReferenceCoverFind
1.
  • Alffenaar, Jan-Willem C., et al. (author)
  • Pharmacokinetics and pharmacodynamics of anti-tuberculosis drugs : An evaluation of in vitro, in vivo methodologies and human studies
  • 2022
  • In: Frontiers in Pharmacology. - : Frontiers Media S.A.. - 1663-9812. ; 13
  • Research review (peer-reviewed)abstract
    • There has been an increased interest in pharmacokinetics and pharmacodynamics (PKPD) of anti-tuberculosis drugs. A better understanding of the relationship between drug exposure, antimicrobial kill and acquired drug resistance is essential not only to optimize current treatment regimens but also to design appropriately dosed regimens with new anti-tuberculosis drugs. Although the interest in PKPD has resulted in an increased number of studies, the actual bench-to-bedside translation is somewhat limited. One of the reasons could be differences in methodologies and outcome assessments that makes it difficult to compare the studies. In this paper we summarize most relevant in vitro, in vivo, in silico and human PKPD studies performed to optimize the drug dose and regimens for treatment of tuberculosis. The in vitro assessment focuses on MIC determination, static time-kill kinetics, and dynamic hollow fibre infection models to investigate acquisition of resistance and killing of Mycobacterium tuberculosis populations in various metabolic states. The in vivo assessment focuses on the various animal models, routes of infection, PK at the site of infection, PD read-outs, biomarkers and differences in treatment outcome evaluation (relapse and death). For human PKPD we focus on early bactericidal activity studies and inclusion of PK and therapeutic drug monitoring in clinical trials. Modelling and simulation approaches that are used to evaluate and link the different data types will be discussed. We also describe the concept of different studies, study design, importance of uniform reporting including microbiological and clinical outcome assessments, and modelling approaches. We aim to encourage researchers to consider methods of assessing and reporting PKPD of anti-tuberculosis drugs when designing studies. This will improve appropriate comparison between studies and accelerate the progress in the field.
  •  
2.
  • Asimus, Sara, 1976, et al. (author)
  • Artemisinin antimalarials moderately affect cytochrome P450 enzyme activity in healthy subjects.
  • 2007
  • In: Fundamental & Clinical Pharmacology. - : Wiley. - 0767-3981 .- 1472-8206. ; 21:3, s. 307-316
  • Journal article (peer-reviewed)abstract
    • The aim of this study was to investigate which principal human cytochrome P450 (CYP450) enzymes are affected by artemisinin and to what degree the artemisinin derivatives differ with respect to their respective induction and inhibition capacity. Seventy-five healthy adults were randomized to receive therapeutic oral doses of artemisinin, dihydroartemisinin, arteether, artemether or artesunate for 5 days (days 1–5). A six-drug cocktail consisting of caffeine, coumarin, mephenytoin, metoprolol, chlorzoxazone and midazolam was administered orally on days −6, 1, 5 and 10 to assess the activities of CYP1A2, CYP2A6, CYP2C19, CYP2D6, CYP2E1 and CYP3A, respectively. Four-hour plasma concentrations of parent drugs and corresponding metabolites and 7-hydroxycoumarin urine concentrations were quantified by liquid chromatography-tandem mass spectrometry. The 1-hydroxymidazolam/midazolam 4-h plasma concentration ratio (CYP3A) was increased on day 5 by artemisinin [2.66-fold (98.75% CI: 2.10–3.36)], artemether [1.54 (1.14–2.09)] and dihydroartemisinin [1.25 (1.06–1.47)] compared with day −6. The S-4'-hydroxymephenytoin/S-mephenytoin ratio (CYP2C19) was increased on day 5 by artemisinin [1.69 (1.47–1.94)] and arteether [1.33 (1.15–1.55)] compared with day −6. The paraxanthine/caffeine ratio (CYP1A2) was decreased on day 1 after administration of artemisinin [0.27 (0.18–0.39)], arteether [0.70 (0.55–0.89)] and dihydroartemisinin [0.73 (0.59–0.90)] compared with day −6. The α-hydroxymetoprolol/metoprolol ratio (CYP2D6) was lower on day 1 compared with day −6 in the artemisinin [0.82 (0.70–0.96)] and dihydroartemisinin [0.83 (0.71–0.96)] groups, respectively. In the artemisinin-treated subjects this decrease was followed by a 1.34-fold (1.14–1.58) increase from day 1 to day 5. These results show that intake of artemisinin antimalarials affect the activities of several principal human drug metabolizing CYP450 enzymes. Even though not significant in all treatment groups, changes in the individual metrics were of the same direction for all the artemisinin drugs, suggesting a class effect that needs to be considered in the development of new artemisinin derivatives and combination treatments of malaria.
  •  
3.
  • Ayoun Alsoud, Rami, et al. (author)
  • Combined quantitative tuberculosis biomarker model for time-to-positivity and colony forming unit to support tuberculosis drug development
  • 2023
  • In: Frontiers in Pharmacology. - : Frontiers Media S.A.. - 1663-9812. ; 14
  • Journal article (peer-reviewed)abstract
    • Biomarkers are quantifiable characteristics of biological processes. In Mycobacterium tuberculosis, common biomarkers used in clinical drug development are colony forming unit (CFU) and time-to-positivity (TTP) from sputum samples. This analysis aimed to develop a combined quantitative tuberculosis biomarker model for CFU and TTP biomarkers for assessing drug efficacy in early bactericidal activity studies. Daily CFU and TTP observations in 83 previously patients with uncomplicated pulmonary tuberculosis after 7 days of different rifampicin monotherapy treatments (10-40 mg/kg) from the HIGHRIF1 study were included in this analysis. The combined quantitative tuberculosis biomarker model employed the Multistate Tuberculosis Pharmacometric model linked to a rifampicin pharmacokinetic model in order to determine drug exposure-response relationships on three bacterial sub-states using both the CFU and TTP data simultaneously. CFU was predicted from the MTP model and TTP was predicted through a time-to-event approach from the TTP model, which was linked to the MTP model through the transfer of all bacterial sub-states in the MTP model to a one bacterial TTP model. The non-linear CFU-TTP relationship over time was well predicted by the final model. The combined quantitative tuberculosis biomarker model provides an efficient approach for assessing drug efficacy informed by both CFU and TTP data in early bactericidal activity studies and to describe the relationship between CFU and TTP over time.
  •  
4.
  • Ayoun Alsoud, Rami, et al. (author)
  • Model-based effect evaluation of a novel Mmpl3 inhibitor in C3HeB/FeJ compared to BALB/c mouse models and translation to humans
  • Other publication (other academic/artistic)abstract
    • Background and Purpose: During tuberculosis drug development, the design of early clinical studies is informed by preclinical animal models. The aim of this work was to describe the exposure-response relationship of a novel inhibitor of mycobacterial MmpL3, prodrug MPL-447, in C3HeB/FeJ mice with non-necrotic or necrotic lesions, and to compare to chronic BALB/c mice information.Experimental Approach: C3HeB/FeJ mice were randomised to placebo and three treatment groups (25, 50 or 100 mg/kg MPL-447). Colony forming unit (CFU) were obtained until week 8 post-treatment. Semi-mechanistic modelling was used to describe growth and killing in relation to exposure. Early bactericidal activity after 14 days (EBA0-14) in humans was predicted using the final model, translational factors and allometric scaling of pharmacokinetics to humans and compared to chronic BALB/c.Key Results: The final model showed 1100% growth and 42% killing of the fast-multiplying bacteria in C3HeB/FeJ mice with necrotic lesions compared to those with non-necrotic lesions. Simulations revealed similar log10CFU reduction on day 14 in C3HeB/FeJ mice with non-necrotic lesions as in chronic BALB/c mice in response to treatment, but 1.7-fold lower reduction in C3HeB/FeJ mice with necrotic lesions. Similar human EBA0-14 was predicted irrespective of the mouse model used. Conclusion and Implications:  The difference in killing of fast-multiplying bacteria in C3HeB/FeJ mice with necrotic lesions compared to C3HeB/FeJ mice without or chronic BALB/C mice was not translated to human early clinical predictions, most likely due to low abundance of these bacteria in humans.
  •  
5.
  • Ayoun Alsoud, Rami, et al. (author)
  • Model-based interspecies scaling for predicting human pharmacokinetics of CB 4332, a complement factor I protein
  • Other publication (other academic/artistic)abstract
    • The extrapolation of a protein pharmacokinetics (PK) from preclinical to clinical studies can be less reliable than for small molecules. CB 4332 is a 150 kDa recombinant complement factor I (CFI) protein. In order to support clinical development, interspecies scaling of CB 4332 using traditional and model-based approaches was performed to inform first-in-human (FIH) dose selection. Plasma concentration versus time data from four preclinical PK studies of single intravenous (i.v.) and subcutaneous (s.c.) CB 4332 dosing in mice, rats and nonhuman primates (NHPs) were modeled simultaneously using naive pooling including allometric scaling. The human-equivalent dose was calculated using the preclinical no observed adverse effect level (NOAEL) as part of the dose-by-factor approach. Pharmacokinetic modelling of CB 4332 revealed species-specific differences in the elimination, which was accounted for by including an additional rat-specific clearance. Signs of anti-drug antibodies (ADA) formation in all rats and some NHPs were observed. Consequently, an additional ADA-induced clearance parameter was estimated including the time of onset. Using the traditional dose-by-factor approach, a maximum recommended starting s.c. dose of 0.9 mg/kg once weekly was calculated using the NOAEL observed in NHPs. The model-based clinical trial simulations predicted it to result in a trough concentration at steady state 12.8% of the determined efficacy target for CB 4332 in humans. Interspecies scaling was performed for CB 4332 using traditional and model-based scaling, where PK modeling allowed the inclusion of preclinical PK information from three species, accounted for potential effects of ADA and species differences in elimination, and allowed the prediction of human PK for FIH dose selection.
  •  
6.
  • Ayoun Alsoud, Rami, 1992- (author)
  • Pharmacometric tools to support translational drug development
  • 2024
  • Doctoral thesis (other academic/artistic)abstract
    • The use of model-informed drug development has been shown to save significant costs and improve decision making early in the drug development process. The work in this PhD thesis aimed to employ pharmacometric tools to support translational drug development from the preclinical to the late clinical stages.Pharmacometric modeling was used to characterize the treatment-shortening potential of different anti tuberculosis regimens. The results provided additional evidence in favor of the treatment-shortening capacity of the BPaMZ regimen over BPaL and standard of care, HRZE.Pharmacokinetic-pharmacodynamic (PKPD) modeling was used to enable the evaluation of the exposure-response of a new anti-tubercular drug, MPL-447, in C3HeB/FeJ mice, thought to be of a translational value in tuberculosis drug development. Model-based evaluation revealed a significant impact of necrotic lesion development in mice on both bacterial growth and sensitivity to treatment with MPL-447, highlighting the significance of accounting for the heterogenous lesion profile in the C3HeB/FeJ mouse model when evaluating drug efficacy.Pharmacokinetic (PK) modeling was employed to perform interspecies PK scaling of the CB 4332 protein using information from three preclinical species. This approach accounted for the impact of immunogenicity and species-related differences in elimination. Simulations predicted the protein plasma concentrations in humans after different dosing regimens and suggested that a 7 mg/kg dose would be required to reach the target at steady-state.Using combined biomarker data, PKPD modeling was employed to simultaneously analyze two tuberculosis efficacy biomarkers. The final biomarker model facilitated the prediction of the relationship between the two biomarkers over time. With this modeling framework, missing biomarker data can be predicted using information from the other biomarker.Several model-based approaches were also explored to evaluate pediatric study power in rare diseases. These approaches were performed analyzing pediatric data alone or combined with the adult data. While Bayesian priors performed well when analyzing pediatric data alone, less technical modeling approaches proved sufficient when pediatric and adult data were combined.In conclusion, the research presented in this thesis has addressed various challenges encountered in translational drug development. The work has contributed to the evaluation of new anti-tubercular drugs and regimens, the assessment of newly proposed animal models, and optimizing the utilization of biomarker information. Furthermore, this thesis has provided insights into the selection of First-in-Human dose for a protein, showcasing the applicability of model-based approaches in this critical decision-making process. The research has contributed to improving analysis approaches for pediatrics in rare diseases.
  •  
7.
  • Björnsson, Marcus A, et al. (author)
  • A Recirculatory Model for Pharmacokinetics and the Effects on Bispectral Index After Intravenous Infusion of the Sedative and Anesthetic AZD3043 in Healthy Volunteers
  • 2015
  • In: Anesthesia and Analgesia. - 0003-2999 .- 1526-7598. ; 121:4, s. 904-913
  • Journal article (peer-reviewed)abstract
    • BACKGROUND: AZD3043 is a positive allosteric modulator of the γ-aminobutyric acid type A receptor, with sedative and anesthetic properties. We describe a population pharmacokinetic (PK) model of arterial and venous concentrations of AZD3043 and the pharmacodynamic effects on bispectral index (BIS) in healthy volunteers.METHODS: Arterial and venous plasma concentrations of AZD3043 and BIS were measured in 2 clinical studies in 125 healthy volunteers, where AZD3043 was given as a 1-minute bolus (1-6 mg/kg), a 30-minute infusion (1-81 mg/kg/h), or 0.8 + 10, 1 + 15, 3 + 30, and 4 + 40 (mg/kg bolus + mg/kg/h infusion for 30 minutes). Population PK/pharmacodynamic analysis was performed with NONMEM.RESULTS: A recirculatory model, comprising a series of 5 compartments for the transit of drug between venous and arterial plasma, 2 peripheral distribution compartments, and 1 compartment for the nondistributive transit of drug from arterial to venous plasma, described the PK of AZD3043. Systemic clearance was high (2.2 L/min; 95% confidence interval, 2.12-2.25), and apparent volumes of distribution were low, leading to a short elimination half-life. The apparent volumes of distribution of the arterial and peripheral compartments increased with increasing administered dose, giving a total apparent volume of distribution of 15 L after the lowest dose and 37 L after the greatest dose. A sigmoid maximum effect (Emax) model with an EC50 of 15.6 µg/mL and a γ of 1.7 described the relationship between AZD3043 effect-site concentrations and BIS. The between-subject variability in EC50 was 37%. An effect compartment model, with a half-life of the equilibration rate constant ke0 of 1.1 min, described the delay in effect in relation to the arterial plasma concentrations.CONCLUSIONS: AZD3043 had a high clearance and a low apparent volume of distribution, leading to a short half-life. However, the apparent volume of distribution was dose dependent (P < 0.001), leading to an increased half-life with increasing dose. The distribution to the effect site was fast and together with the short plasma half-life led to a fast onset and offset of effects.
  •  
8.
  • Björnsson, Marcus A., et al. (author)
  • A two-compartment effect site model describes the bispectral index after different rates of propofol infusion
  • 2010
  • In: Journal of Pharmacokinetics and Pharmacodynamics. - : Springer Science and Business Media LLC. - 1567-567X .- 1573-8744. ; 37:3, s. 243-255
  • Journal article (peer-reviewed)abstract
    • Different estimates of the rate constant for the effect site distribution (k(e0)) of propofol, depending on the rate and duration of administration, have been reported. This analysis aimed at finding a more general pharmacodynamic model that could be used when the rate of administration is changed during the treatment. In a cross-over study, 21 healthy volunteers were randomised to receive a 1 min infusion of 2 mg/kg of propofol at one occasion, and a 1 min infusion of 2 mg/kg of propofol immediately followed by a 29 min infusion of 12 mg kg(-1) h(-1) of propofol at another occasion. Arterial plasma concentrations of propofol were collected up to 4 h after dosing, and BIS was collected before start of infusion and until the subjects were fully awake. The population pharmacokinetic-pharmacodynamic analysis was performed using NONMEM VI. A four-compartment PK model with time-dependent elimination and distribution described the arterial propofol concentrations, and was used as input to the pharmacodynamic model. A standard effect compartment model could not accurately describe the delay in the effects of propofol for both regimens, whereas a two-compartment effect site model significantly improved the predictions. The two-compartment effect site model included a central and a peripheral effect site compartment, possibly representing a distribution within the brain, where the decrease in BIS was linked to the central effect site compartment concentrations through a sigmoidal E-max model.
  •  
9.
  • Björnsson, Marcus A., et al. (author)
  • Modelling of pain intensity and informative dropout in a dental pain model after naproxcinod, naproxen and placebo administration
  • 2011
  • In: British Journal of Clinical Pharmacology. - : Wiley. - 0306-5251 .- 1365-2125. ; 71:6, s. 899-906
  • Journal article (peer-reviewed)abstract
    • AIMS To describe pain intensity (PI) measured on a visual analogue scale (VAS) and dropout due to request for rescue medication after administration of naproxcinod, naproxen or placebo in 242 patients after wisdom tooth removal. METHODS Non-linear mixed effects modelling was used to describe the plasma concentrations of naproxen, either formed from naproxcinod or from naproxen itself, and their relationship to PI and dropout. Goodness of fit was assessed by simultaneous simulations of PI and dropout. RESULTS Baseline PI for the typical patient was 52.7 mm. The PI was influenced by placebo effects, using an exponential model, and by naproxen concentrations using a sigmoid E-max model. Typical maximal placebo effect was a decrease in PI by 20.2%, with an onset rate constant of 0.237 h-1. EC50 was 0.135 mu mol l-1. A Weibull time-to-event model was used for the dropout, where the hazard was dependent on the predicted PI and by the PI at baseline. Since the dropout was not at random, it was necessary to include the simulated dropout in visual predictive checks (VPC) of PI. CONCLUSIONS This model describes the relationship between drug effects, PI and the likelihood of dropout after naproxcinod, naproxen and placebo administration. The model provides an opportunity to describe the effects of other doses or formulations, after dental extraction. VPC created by simultaneous simulations of PI and dropout provides a good way of assessing the goodness of fit when there is informative dropout.
  •  
10.
  • Boström, Emma, et al. (author)
  • Blood–Brain Barrier Transport Helps to Explain Discrepancies in In Vivo Potency between Oxycodone and Morphine
  • 2008
  • In: Anesthesiology. - 0003-3022 .- 1528-1175. ; 108:3, s. 495-505
  • Journal article (peer-reviewed)abstract
    • Background The objective of this study was to evaluate the brain pharmacokinetic-pharmacodynamic relations of un-bound oxycodone and morphine to investigate the influence of blood-brain barrier transport on differences in potency between these drugs. Methods: Microdialysis was used to obtain unbound concentrations in brain and blood. The antinociceptive effect of each drug was assessed using the hot water tail-flick method. Population pharmacokinetic modeling was used to describe the bloodbrain barrier transport of morphine as the rate (Cl.) and extent (K-p,K-uu) of equilibration, where CLin is the influx clearance across the blood-brain barrier and Kp,,,, is the ratio of the unbound concentration in brain to that in blood at steady state. Results: The six-fold difference in K-p,K-uu between oxycodone and morphine implies that, for the same unbound concentration in blood, the concentrations of unbound oxycodone in brain will be six times higher than those of morphine. A joint pharmacokinetic-pharmacodynamic model of oxycodone and morphine based on unbound brain concentrations was developed and used as a statistical tool to evaluate differences in the pharmacodynamic parameters of the drugs. A power model using Effect = Baseline + Slope center dot C-gamma best described the data. Drug-specific slope and gamma parameters made the relative potency of the drugs concentration dependent. Conclusions: For centrally acting drugs such as opioids, pharmacokinetic-pharmacodynamic relations describing the interaction with the receptor are better obtained by correlating the effects to concentrations of unbound drug in the tissue of interest rather than to blood concentrations.
  •  
Skapa referenser, mejla, bekava och länka
  • Result 1-10 of 113
Type of publication
journal article (99)
other publication (5)
doctoral thesis (5)
research review (4)
Type of content
peer-reviewed (98)
other academic/artistic (15)
Author/Editor
Simonsson, Ulrika S. ... (70)
Simonsson, Ulrika S. ... (43)
Svensson, Robin J. (14)
Diacon, Andreas H. (13)
Karlsson, Mats O. (13)
Faraj, Alan (13)
show more...
Van Wijk, Rob C, 199 ... (12)
McIlleron, Helen (10)
Wicha, Sebastian G. (9)
Clewe, Oskar (9)
Keutzer, Lina (8)
Svensson, Elin, 1985 ... (7)
Kreilgaard, Mads (7)
Gillespie, Stephen H ... (7)
Upton, Caryn M. (7)
de Steenwinkel, Jurr ... (6)
Nyberg, Joakim, 1978 ... (6)
Ayoun Alsoud, Rami (6)
Knudsen, Tom (6)
Svensson, Elin M., 1 ... (5)
Smith, Peter (5)
Ashton, Michael, 195 ... (5)
Jansson, Britt (5)
Boeree, Martin J. (5)
Niward, Katarina (5)
Boström, Emma (4)
Dawson, Rodney (4)
Denti, Paolo (4)
Jönsson, Siv (4)
Hammarlund-Udenaes, ... (4)
Christrup, Lona Lour ... (4)
Krekels, Elke H. J. (4)
Hu, Yanmin (4)
Coates, Anthony R M (4)
McIlleron, Helen M. (4)
Smith, Peter J (4)
Schön, Thomas (3)
Lennernäs, Hans (3)
Miller, Raymond (3)
Bruchfeld, Judith (3)
Elsherbiny, Doaa (3)
Eliasson, Erik (3)
Björnsson, Marcus A (3)
Chen, Chunli (3)
van der Walt, Jan-St ... (3)
Karlsson, Mats O., P ... (3)
Wilkins, Justin J. (3)
Aarnoutse, Rob (3)
Blouse, Grant E. (3)
Paues, Jakob, 1972- (3)
show less...
University
Uppsala University (113)
Karolinska Institutet (10)
University of Gothenburg (6)
Linköping University (5)
Lund University (1)
Language
English (113)
Research subject (UKÄ/SCB)
Medical and Health Sciences (106)
Natural sciences (1)
Agricultural Sciences (1)

Year

Kungliga biblioteket hanterar dina personuppgifter i enlighet med EU:s dataskyddsförordning (2018), GDPR. Läs mer om hur det funkar här.
Så här hanterar KB dina uppgifter vid användning av denna tjänst.

 
pil uppåt Close

Copy and save the link in order to return to this view